You are on page 1of 6

Targeted

cancer nanotherapy
by Gloria J. Kim and Shuming Nie*

Significant progress has been made in the Conventional cancer therapy and diagnostics involves
development of new agents against cancer and new the application of catheters, surgery, biopsy,
chemotherapy, and radiation. Most current anticancer
delivery technologies. Proteomics and genomics
agents do not greatly differentiate between
continue to uncover molecular signatures that are
cancerous and normal cells. This leads to systemic
unique to cancer. Yet, the major challenge remains in toxicity and adverse effects. Consequently, the
targeting and selectively killing cancer cells while systemic application of these drugs often causes
affecting as few healthy cells as possible. severe sideeffects in other tissues (e.g. bone marrow
Nanometer-sized particles have novel optical, suppression, cardiomyopathy, and neurotoxicity),
electronic, and structural properties that are not which greatly limits the maximal allowable dose of
available from either individual molecules or bulk the drug. In addition, rapid elimination and
widespread distribution into nontargeted organs and
solids. When linked with tumor-targeting moieties,
tissues requires the administration of a drug in large
such as tumor-specific ligands or monoclonal
quantities, which is uneconomical and is often
antibodies, these nanoparticles can be used to target complicated because of nonspecific toxicity.
cancer-specific receptors, tumor antigens Nanotechnology could offer a less invasive alternative,
(biomarkers), and tumor vasculatures with high enhancing the life expectancy and quality of life of the
affinity and precision. patient. The diameter of human cells spans 10-20 µm. The size
of cell organelles ranges from a few nanometers to a few
hundred nanometers. Nanoscale devices can readily interact
with biomolecules on the cell surface and within the cells in
a noninvasive manner, leaving the behavior and biochemical
properties of those molecules intact. In their ‘mesoscopic’
size range of 10-100 nm in diameter, nanoparticles have
more surface areas and functional groups that can be linked
to multiple optical, radioisotopic, or magnetic diagnostic and
therapeutic agents. When linked with tumor-targeting
Department of Biomedical Engineering, ligands such as monoclonal antibodies, these nanoparticles
Emory University and Georgia Institute of Technology,
101 Woodruff Circle Suite 2001, can be used to target tumor antigens (biomarkers), as well as
Atlanta, GA 30322, USA
*E-mail: snie@emory.edu
tumor vasculatures with high affinity and specificity. In this

28 August 2005 ISSN:1369 7021 © Elsevier Ltd 2005


RESEARCH REPORT
Glossary
Passive Targeting EPR Effect
Agonist A drug that triggers an action from a cell or
another drug.
Tumor Environment
Angiogenesis The process of vascularization of a
tissue involving the development of new capillary blood Direct Local Delivery

vessels. This is caused by the release of chemicals by


Active Targeting Lectin-carbohydrate Interaction
the tumor.

Antagonist A compound that inhibits the effect of a Ligand-receptor Interaction


hormone or drug.
Antibody-antigen Interaction
Apoptosis A form of programmed cell death induced
by external or internal signals that trigger the activity Scheme 1. Targeting strategies for nanoscale drug delivery systems.
of proteolytic caspases, whose actions dismantle the
article, we discuss different targeting strategies for nanoscale
cell and result in cell death.
drug delivery systems (see Scheme 1), and offer a perspective
Doxorubicin A type of anticancer drug called an on cancer nanotherapy.
‘anthracycline glycoside’. It works by impairing DNA
synthesis, and hence can target rapidly dividing cells. Passive targeting
Solid tumors have a diffusion-limited maximal size1,2 of
Epitope A part of a molecule that an antibody will
about 2 mm3 and will remain at this size until angiogenesis
recognize and bind to.
occurs, thus granting them access to the circulation3. Rapid
Integrin Superfamily of cell surface proteins that are vascularization to serve fast-growing cancerous tissues
involved in binding to extracellular matrix components inevitably leads to a leaky, defective architecture and
in some cases. impaired lymphatic drainage. This structure allows an
enhanced permeation and retention (EPR) effect4,5 (first
Liposome Lipid bilayers that form colloidal particles in
described by Matsumura et al.6), as a result of which
an aqueous medium.
nanoparticles accumulate at the tumor site. For such a
Matrix metalloproteinase A member of a group of passive targeting mechanism to work, the size and surface
enzymes that can break down proteins, such as properties of drug delivery nanoparticles must be controlled
collagen, normally found in the spaces between cells to avoid uptake by the reticuloendothelial system (RES)7. To
in tissues (i.e. extracellular matrix proteins). They maximize circulation times and targeting ability, the optimal
need Zn or Ca ions to work properly. Matrix size should be less than 100 nm in diameter and the surface
metalloproteinases are involved in wound healing, should be hydrophilic to circumvent clearance by
angiogenesis, and tumor cell metastasis. macrophages (large phagocytic cells of the RES). A
hydrophilic nanoparticle surface safeguards against plasma
Reticuloendothelial system A system composed of protein adsorption, and can be achieved through hydrophilic
monocytes and macrophages located in reticular polymer coating (e.g. by polyethylene glycol (PEG),
connective tissue. These cells are responsible for poloxamines, poloxamers, and polysaccharides) or the use of
engulfing (phagocytosis) and removing cellular debris, branched or block copolymers8,9. The covalent linkage of
old cells, pathogens, and foreign substances from the amphiphilic copolymers (polylactic acid, polycaprolactone,
bloodstream. and polycyanonacrylate) chemically coupled to PEG9-11 is
generally preferred, as it avoids aggregation and ligand
Xenobiotics Chemicals that are foreign to the body.
desorption when in contact with blood components7.
Xenograft Cells of one species transplanted to another. An alternative passive targeting strategy is to use the
unique tumor environment in a scheme called tumor-activated

August 2005 29
RESEARCH REPORT

Biocompatible polymer
Lectin-carbohydrate is one of the classic examples for
Anticancer
agent targeted drug delivery15. Lectins are proteins of
nonimmunological origin that are capable of recognizing and
binding to glycoproteins expressed on cell surfaces. Lectin
Cleavable linkage
responsive to tumor environment interactions with certain carbohydrates are very specific.
Carbohydrate moieties can be used to target drug delivery
systems to lectins (direct lectin targeting), and lectins can be
used as targeting moieties to target cell surface
carbohydrates (reverse lectin targeting). However, drug
delivery systems based on lectin-carbohydrate have been
nucleus
developed mainly to target whole organs16, which can harm
normal cells. Therefore, in most cases, the targeting moiety is
Fig. 1 Tumor-activated prodrug delivery and targeting. The anticancer agent is conjugated directed toward specific receptors or antigens expressed on
to a biocompatible polymer via an ester bond. The linkage is hydrolyzed by cancer-specific
enzymes, or by high or low pH, at the tumor site, at which time the nanoparticle releases the plasma membrane or elsewhere at the tumor site.
the drug.
Multiple drug resistance (MDR), which is a major challenge
prodrug therapy. The drug is conjugated to a tumor-specific in chemotherapy, often stems from the overexpression of the
molecule and remains inactive until it reaches the target12 plasma membrane P-glycoprotein (Pgp)17,18. In general, Pgp
(Fig. 1). Overexpression of the matrix metalloproteinase acts as an efflux pump to extrude positively charged
(MMP), MMP-2, in melanoma has been shown in a number of xenobiotics – including some anticancer drugs – out of the
preclinical as well as clinical investigations. Mansour et al.13 cell. Many tumor cells are resistant to doxorubicin, which is a
reported a water-soluble maleimide derivative of doxorubicin Pgp substrate. To overcome the resistance,
(DOX) incorporating an MMP-2-specific peptide sequence poly(cyanocarylate) nanoparticles have been developed19,20.
(Gly-Pro-Leu-Gly-Ile-Ala-Gly-Gln) that binds rapidly and Adsorption of the nanoparticles onto the plasma membrane
selectively to the cysteine-34 position of circulating albumin. and the subsequent release of doxorubicin leads to saturation
The albumin-doxorubicin conjugate is cleaved efficiently and of Pgp. Furthermore, the negatively charged degradation
specifically by MMP-2, releasing a doxorubicin tetrapeptide products of the polymer form an ion pair and neutralize the
(Ile-Ala-Gly-Gln-DOX) and subsequently doxorubicin. pH and positive charge of doxorubicin19, enhancing the diffusion of
redox potential have also been explored as drug-release the drug across the plasma membrane. Blagosklonny
triggers at the tumor site14. proposed an approach to selectively kill resistant cancer cells
Yet another passive targeting method is the direct local that is based on a temporary increase in the resistance of
delivery of anticancer agents to tumors. This approach has sensitive cells against certain drugs by specific protectors,
the obvious advantage of excluding the drug from the such as pharmacological inhibitors of apoptosis21. These
systemic circulation. However, administration can be highly protectors are pumped out by MDR cells, while increasing the
invasive, as it involves injections or surgical procedures. For resistance in sensitive cells that do not have active drug
some tumors that are difficult to access, such as lung cancers, efflux pumps. After applying a cytotoxic drug, sensitive cells
the technique is nearly impossible to use. are protected and survive the exposure, while unprotected
MDR counterparts are killed. By abolishing dose-limiting side-
Active targeting effects of chemotherapy, this strategy might provide a means
Active targeting is usually achieved by conjugating to the to selectively treat aggressive and resistant cancers. Tsuruo
nanoparticle a targeting component that provides preferential suggested that antibodies to P-glycoprotein overexpressed on
accumulation of nanoparticles in the tumor-bearing organ, in multidrug resistant (MDR) cells could make an attractive
the tumor itself, individual cancer cells, intracellular targeting moiety22.
organelles, or specific molecules in cancer cells. This approach The overexpression of receptors or antigens in many
is based on specific interactions such as lectin-carbohydrate, human cancers lends itself to efficient drug uptake via
ligand-receptor, and antibody-antigen. receptor-mediated endocytosis (cellular ingestion) – see Fig. 2.

30 August 2005
RESEARCH REPORT

Since glycoproteins cannot remove polymer-drug conjugates Cargo

that have entered the cells via endocytosis23,24, this active Ligand

targeting mechanism provides an alternative route for Cleavable Bond

overcoming MDR. Receptor

The cell surface receptor for folate is inaccessible from the 1. Internalization

circulation to healthy cells because of its location on the apical Cancer Cell Surface
H+
H+
membrane of polarized epithelia, but is overexpressed on the
6. Membrane Fusion
surface of various cancers, including ovary, brain, kidney, H+

breast, and lung malignancies. Surface plasmon resonance


2. Early Endosomes H+
studies reveal that folate-conjugated PEGylated cyanoacrylate H+
5. Recycling Endosomes
nanoparticles have a ten-fold higher affinity for the folate
H+
H+ H+
receptor than free folate11. Folate receptors are often H+

organized in clusters and bind preferably to the multivalent 3. Acidified Endosomes 4. Endosomal Release

forms of the ligand. Furthermore, confocal microscopy


Fig. 2 Nanoparticle drug delivery and targeting using receptor-mediated endocytosis.
demonstrated selective uptake and endocytosis of folate-
conjugated nanoparticles by tumor cells that bear folate with better success. For example, anti-ERBB2
receptors. Interest in exploiting folate receptor targeting in immunoliposomes loaded with doxorubicin show greater
cancer therapy and diagnosis has increased rapidly, as attested antitumor activity than the free drug or the drug loaded in
by many conjugated systems, including proteins, liposomes, nontargeted liposomes in several tumor xenograft
imaging agents, and neutron activation compounds9,11,25-31. models39,40. Moreover, the systemic toxicity of the
Tumor targeting by antibodies with engineered properties immunoliposome-targeted doxorubicin was much less than
(Table 1) is in its infancy, but holds much promise. The that of free doxorubicin. Bispecific antibodies, which are non-
monoclonal antibody (mAb) BR96 (anti-sialyl Lewis Y natural antibodies with two different epitopes, have been
antigen), conjugated with doxorubicin, has proved highly used most widely for the delivery of immune effector cells
efficacious in tumour xenograft studies32 but, unfortunately, and, to a lesser extent, for the delivery of radionuclides,
has shown little or no efficacy in Phase II trials for metastatic drugs, and toxins to tumors41,42.
breast cancer33 and advanced gastric adenocarcinoma34, Alternatively, tumor vasculatures can be targeted to allow
respectively. Moreover, dose-limiting gastrointestinal targeted delivery to a wide range of tumor types. A number of
toxicities are observed in the breast cancer trial, because the angiogenesis inhibitors are undergoing clinical trials (Table 2).
immunoconjugate binds to antigen-positive normal cells in Antiangiogenic therapy prevents neovascularization (the
gastric mucosa, small intestine, and pancreas33. proliferation of blood vessels in different tissues) by
Calicheamicins35-37 and maytansinoids38 are the most inhibiting proliferation, migration, and differentiation of
extensively evaluated of many small-molecule toxins that are endothelial cells43. Vascular endothelial growth factor (VEGF)
used for direct antibody arming, but indirect arming has met is expressed in many solid tumors. A potent angiogenesis-

Table 1 Antibodies in cancer therapeutics.

Mechanism Antibody target Trade name


Agonist activity CD40, CD137 Various
Antagonist activity CTLA4 MDX-010
Angiogenesis inhibition VEGF Avastin™
Antibody-dependent cell-mediated cytotoxicity CD20 Rituxan®, HuMax-CD20
HER-2/neu Herceptin®
EGF receptor HuMax-EGFr
Toxin-mediated killing CD33 Mylotarg®
Disruption signaling HER-2/neu Pertuzumab (2C4)
Complement-dependent cytotoxicity CD20 Rituxan®, HuMax-CD20
Blockage ligand binding EGF receptor Erbitux™

August 2005 31
RESEARCH REPORT

Table 2 Angiogenesis inhibitors undergoing clinical trial.

Mechanism Antiangiogenic drug Target Stage of clinical development


Monoclonal antibodies targeting VEGF-A Avastin™ VEGF-A Phase I, II, III
VEGF-Trap VEGF-A Phase I
Antibodies targeting VEGFR-2 IMC-ICII VEGFR-2 Phase I
Receptor tyrosine kinase inhibitors SU5416 VEGFR-2 Phase I, II, III
SU6668 VEGFR-2, bFGFR, PDGFR Phase I, II
ZD 6474 VEGFR-2, EGFR Phase I, II
Endothelial cell proliferation inhibitors ABT-510 Endothelial CD-36 Phase I, II
Angiostatin Various Phase I
Endostatin Various Phase I
TNP-470 Methionine aminopeptidase, Phase I
cyclin dependent kinase 2
Integrin activity inhibitors Vitaxin Integrin ανβ3 Phase I, II
Medi-522 Integrin αvβ3 Phase I
Vascular targeting agents ZD 6126 Endothelial tubulin Phase I
DMXAA Endothelial tubulin Phase I

stimulating protein, it also increases the permeability of nonionizable compounds, micronization, soft-gel technology,
tumor blood vessels. This leads to swelling of the tumor, cosolvents, surfactants, or complexing agents have been
ultimately hindering the ability of cancer cells to recruit used52,34. Since it is faster and more cost effective to
blood supply through angiogenesis. VEGF has been used in redesign the molecule than to develop a new one, a broadly
liposomes and polymeric nanospheres to deliver angiostatin based technology applicable to poorly water-soluble drugs
and endostatin44,45. The αvβ3 integrin is one of the most could have a tremendous impact.
specific biomarkers that can differentiate newly formed Paclitaxel (Taxol™) is a microtubule-stabilizing agent that
capillaries from their mature counterparts46. Although all promotes tubulin polymerization, disrupting cell division and
endothelial cells use integrin receptors to attach to the causing cell death54. It displays neoplastic activity against
extraluminal submatrix, one unique receptor (αvβ3 integrin) primary epithelial ovarian carcinoma, breast, colon, and lung
is found on the luminal surface of the endothelial cell only cancers. Because it is poorly soluble in aqueous solution, the
during angiogenesis47. High-affinity αvβ3 selective ligands, formulation that is currently available is Chremophor EL
Arg-Gly-Asp (RGD), have been identified by phage display (polyethoxylated castor oil) and ethanol55. In a new
studies. The cyclic form, which contains a conformationally formulation used in Abraxane (recently approved by the FDA
constrained RGD, has a higher binding affinity than the linear to treat metastatic breast cancer), paclitaxel was conjugated
form48. Doxorubicin-loaded PEG nanoparticles conjugated to
cyclic RGD49 and paclitaxel-cyclic RGD nanoparticles50 have Targeting ligand Self-assembled polymer

been reported recently. Anticancer


agent

Covalent bond Tumor specific

Perspective enzyme cleavable linkage

100 nm Receptor-mediated
Nanotechnology is beginning to change the scale and endocytosis

methods of drug delivery. For decades, researchers have been


developing new anticancer agents and new formulations for
delivering existing and new agents.
More than 40% of active substances identified through nucleus

combinatorial screening programs are poorly soluble in Solid tumor

water51. The conventional, and most current, formulations of Receptor nucleus


Release of the anticancer agent
such drugs are frequently plagued with problems such as poor
and inconsistent bioavailability. The most widely used Fig. 3 Self-assembled polymeric nanoparticles for both tumor targeting and therapeutic
functions. Inset: delivery of the nanoparticle drugs by receptor-mediated endocytosis and
method for enhancing solubility is to generate a salt. For controlled drug release inside the cytoplasm.

32 August 2005
RESEARCH REPORT

to albumin nanoparticles56. In addition to circumventing side- Self-assembled polymer

effects of the highly toxic Chremophor EL, which include Anticancer agent
Imaging agent
hypersensitivity reactions and toxicity to kidney cells and
nerve tissue (nephrotoxicity and neurotoxicity)55,57, the
system cleverly takes advantage of albumin receptors for Tumor specific
enzyme cleavable linkage
improved drug delivery to cancer cells.
Covalent bond
For specific targeting, the differences between cancerous
100 nm Targeting ligand 100 nm
cells and normal cells, which include uncontrolled
proliferation, insensitivity to negative growth regulation, and Fig. 4 Multifunctional nanoparticles for integrated cancer imaging and therapy. As
antigrowth signals, angiogenesis and metastasis can be nanoparticles are developed for clinical applications, an exciting opportunity is to monitor
drug delivery and treatment efficacy by using embedded imaging agents.
exploited. Thanks to recent advances in proteomics and
genomics, there is a growing body of knowledge of unique biomedical applications. Many of the principles used to target
cancer markers. They form the basis of complex interactions delivery of drugs to cancers may also be applied to target
between bioconjugated nanoparticles and cancer cells. Carrier imaging and diagnostic agents. The full in vivo potential of
design and targeting strategies may vary according to the cancer nanotechnology in targeted drug delivery and imaging
type, developmental stage, and location of cancer. There is can be realized by using strategically engineered
much synergy between imaging and nanotechnology in multifunctional nanoparticles (Figs. 3 and 4). NT

REFERENCES 29. Leamon, C. P., and Low, P. S., J. Drug Target. (1994) 2 (2), 101
1. Grossfeld, G. D., et al., Urology (2002) 59 (1), 97 30. Konda, S. D., et al., MAGMA (2001) 12 (2-3) 104
2. Storm, G., et al., Adv Drug Deliv Rev. (1995) 17 (1), 31 31. Goren, D., et al., Clin. Cancer Res. (2000) 6 (5), 1949
3. Jain, R. K., Annu. Rev Biomed Eng. (1999) 1, 241 32. Trail, P. A., et al., Science (1993) 261, 212
4. Teicher, B. A., Drug Resist. Update (2000) 3 (2), 67 33. Tolcher, A. W., et al., J. Clin. Oncol. (1999) 17 (2), 478
5. Duncan, R., Nat. Rev. Drug Discov. (2003) 2 (5), 347 34. Ajani, J. A., et al., Cancer J. (2000) 6, 78
6. Matsumura, Y., and Maeda, H., Cancer Res. (1986) 46 (12), 6387 35. Lode, H. N., et al., Cancer Res. (1998) 58 (14), 2925
7. Gref, R., et al., Science (1994) 263, 1600 36. Hinman, L. M., et al., Cancer Res. (1993) 53 (14), 3336
8. Moghimi, S. M., and Hunter, A. C., Trends Biotechnol. (2000) 18 (10), 412 37. Sievers, E. L., et al., J. Clin. Oncol. (2001) 19 (13), 3244
9. Park, E. K., et al., Biomaterials (2005) 26 (9), 1053 38. Liu, C., et al., Proc. Natl. Acad. Sci. U.S.A. (1996) 93 (16), 8618
10. Farokhzad, O. C., et al., Cancer Res. (2004) 64 (21), 7668 39. Park, J. W., et al., Adv. Pharmacol. (1997) 40, 399
11. Stella, B., et al., J. Pharm Sci. (2000) 89 (11), 1452 40. Park, J. W., et al., Cancer Lett. (1997) 118 (2), 153
12. Chari, R. V., Adv. Drug Delivery Rev. (1998) 31 (1-2), 89 41. Koelemij, R., et al., J. Immunother. (1999) 22 (6), 514
13. Mansour, A. M., et al., Cancer Res. (2003) 63 (14), 4062 42. Segal, D. M., et al., Curr. Opin. Immunol. (1999) 11 (5), 558
14. Guo, X.and Szoka, F. C., Jr., Acc. Chem. Res. (2003) 36 (5), 335 43. Jain, R. K., Science (2005) 307, 58
15. Kannagi, R., et al., Cancer Sci. (2004) 95 (5), 377 44. Chen, Q. R., et al., Mol. Genet. Metab. (2001) 74, 120
16. Yamazaki, N., et al., Adv. Drug Delivery Rev. (2000) 43 (2-3), 225 45. Reynolds, A. R., et al., Trends Mol. Med. (2003) 9 (1), 2
17. Links, M.and Brown, R., Expert Rev. Mol. Med. (1999) 1999, 1 46. Brooks, P. C., et al., Cell (1994) 79 (7), 1157
18. Krishna, R.and Mayer, L. D., Eur. J. Pharm. Sci. (2000) 11 (4), 265 47. Cleaver, O., and Melton, D. A., Nat. Med. (2003) 9 (6), 661
19. Vauthier, C., et al., J. Controlled Release (2003) 93 (2), 151 48. Cheng, S., et al., J. Med. Chem. (1994) 37 (1), 1
20. de Verdiere, A. C., et al., Br. J. Cancer (1997) 76 (2), 198 49. Bibby, D. C., et al., Int. J. Pharm. (2005) 293 (1-2) 281
21. Blagosklonny, M. V., Trends Mol. Med. (2003) 9 (7), 307 50. Chen, X., et al., J. Med. Chem. (2005) 48 (4), 1098
22. Tsuruo, T., Intern Med. (2003) 42 (3), 237 51. Merisko-Liversidge, E., et al., Eur. J. Pharm. Sci. (2003) 18 (2), 113
23. Bennis, S., et al., Eur. J. Cancer (1994) 30A (1), 89 52. Fishman, M. L., et al., Biomacromolecules (2004) 5 (2), 334
24. Larsen, A. K., et al., Pharmacol. Ther. (2000) 85 (3), 217 53. Stella, V. J., and Rajewski, R. A., Pharm. Res. (1997) 14 (5), 556
25. Xu, L., et al., J. Controlled Release (2001) 74 (1-3), 115 54. Diaz, J. F., et al., J. Biol. Chem. (2000) 275 (34), 26265
26. Wang, S., et al., Proc. Natl. Acad. Sci. U.S.A. (1995) 92 (8), 3318 55. Singla, A. K., et al., Int. J. Pharm. (2002) 235 (1-2), 179
27. Shukla, S., et al., Bioconjugate Chem. (2003) 14 (1), 158 56. Garber, K., J. Natl. Cancer Inst. (2004) 96 (2), 90
28. Moon, W. K., et al., Bioconjugate Chem. (2003) 14 (3), 539 57. Gelderblom, H., et al., Eur. J. Cancer (2001) 37 (13), 1590

August 2005 33

You might also like